Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Infect Immun ; 88(7)2020 06 22.
Article in English | MEDLINE | ID: mdl-32284374

ABSTRACT

Recent studies have determined that inflammasome signaling plays an important role in driving intestinal epithelial cell (IEC) responses to bacterial infections, such as Salmonella enterica serovar Typhimurium. There are two primary inflammasome pathways, canonical (involving caspase-1) and noncanonical (involving caspase-4 and -5 in humans and caspase-11 in mice). Prior studies identified the canonical inflammasome as the major pathway leading to interleukin-18 (IL-18) release and restriction of S Typhimurium replication in the mouse cecum. In contrast, the human C2Bbe1 colorectal carcinoma cell line expresses little caspase-1 but instead utilizes caspase-4 to respond to S Typhimurium infection. Intestinal enteroid culture has enabled long-term propagation of untransformed IECs from multiple species, including mouse and human. Capitalizing on this technology, we used a genetic approach to directly compare the relative importance of different inflammatory caspases in untransformed mouse and human IECs and transformed human IECs upon S Typhimurium infection in vitro We show that caspase-1 is important for restricting intracellular S Typhimurium replication and initiating IL-18 secretion in mouse IECs but is dispensable in human IECs. In contrast, restriction of intracellular S Typhimurium and production of IL-18 are dependent on caspase-4 in both transformed and untransformed human IECs. Notably, cytosolic replication in untransformed cells from both species was less pronounced than in transformed human cells, suggesting that transformation may impact additional pathways that restrict S Typhimurium replication. Taken together, these data highlight the differences between mouse and human IECs and the utility of studying transformed and untransformed cells in parallel.


Subject(s)
Inflammasomes/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Salmonella Infections/metabolism , Salmonella Infections/microbiology , Salmonella enterica/physiology , Animals , Biomarkers , Caspases/metabolism , Cell Line , Cytokines/metabolism , Disease Models, Animal , Gene Expression , Humans , Intestinal Mucosa/pathology , Mice , Salmonella Infections/genetics
2.
Viruses ; 10(5)2018 04 26.
Article in English | MEDLINE | ID: mdl-29701691

ABSTRACT

Paneth cells are major secretory cells located in the crypts of Lieberkühn in the small intestine. Our understanding of the diverse roles that Paneth cells play in homeostasis and disease has grown substantially since their discovery over a hundred years ago. Classically, Paneth cells have been characterized as a significant source of antimicrobial peptides and proteins important in host defense and shaping the composition of the commensal microbiota. More recently, Paneth cells have been shown to supply key developmental and homeostatic signals to intestinal stem cells in the crypt base. Paneth cell dysfunction leading to dysbiosis and a compromised epithelial barrier have been implicated in the etiology of Crohn’s disease and susceptibility to enteric bacterial infection. Our understanding of the impact of Paneth cells on viral infection is incomplete. Enteric α-defensins, produced by Paneth cells, can directly alter viral infection. In addition, α-defensins and other antimicrobial Paneth cell products may modulate viral infection indirectly by impacting the microbiome. Here, we discuss recent insights into Paneth cell biology, models to study their function, and the impact, both direct and indirect, of Paneth cells on enteric viral infection.


Subject(s)
Intestinal Diseases/pathology , Paneth Cells/metabolism , Virus Diseases/pathology , Animals , Antimicrobial Cationic Peptides/metabolism , Humans , Intestinal Diseases/metabolism , Intestinal Diseases/virology , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Microbiota/physiology , Organoids/metabolism , Organoids/pathology , Organoids/virology , Paneth Cells/cytology , Protein Processing, Post-Translational , Virus Diseases/metabolism
3.
J Virol ; 92(9)2018 05 01.
Article in English | MEDLINE | ID: mdl-29467318

ABSTRACT

Human adenoviruses (HAdV) are significant human pathogens. Although only a subset of HAdV serotypes commonly cause gastroenteritis in humans, most HAdV species replicate in the gastrointestinal tract. Knowledge of the complex interaction between HAdVs and the human intestinal epithelium has been limited by the lack of a suitable cell culture system containing relevant cell types. Recently, this need has been met by the stable and prolonged cultivation of primary intestinal epithelial cells as enteroids. Human enteroids have been used to reveal novel and interesting aspects of rotavirus, norovirus, and enterovirus replication, prompting us to explore their suitability for HAdV culture. We found that both prototype strains and clinical isolates of enteric and nonenteric HAdVs productively replicate in human enteroids. HAdV-5p, a respiratory pathogen, and HAdV-41p, an enteric pathogen, are both sensitive to type I and III interferons in human enteroid monolayers but not A549 cells. Interestingly, HAdV-5p, but not HAdV-41p, preferentially infected goblet cells. And, HAdV-5p but not HAdV-41p was potently neutralized by the enteric human alpha-defensin HD5. These studies highlight new facets of HAdV biology that are uniquely revealed by primary intestinal epithelial cell culture.IMPORTANCE Enteric adenoviruses are a significant cause of childhood gastroenteritis worldwide, yet our understanding of their unique biology is limited. Here we report robust replication of both prototype and clinical isolates of enteric and respiratory human adenoviruses in enteroids, a primary intestinal cell culture system. Recent studies have shown that other fastidious enteric viruses replicate in human enteroids. Therefore, human enteroids may provide a unified platform for culturing enteric viruses, potentially enabling isolation of a greater diversity of viruses from patients. Moreover, both the ability of interferon to restrict respiratory and enteric adenoviruses and a surprising preference of a respiratory serotype for goblet cells demonstrate the power of this culture system to uncover aspects of adenovirus biology that were previously unattainable with standard cell lines.


Subject(s)
Adenovirus Infections, Human/virology , Adenoviruses, Human/isolation & purification , Goblet Cells/virology , Intestine, Small/virology , Virus Replication , Adenovirus Infections, Human/drug therapy , Adenovirus Infections, Human/immunology , Adult , Antiviral Agents/pharmacology , Goblet Cells/drug effects , Goblet Cells/immunology , Humans , Interferons/pharmacology , Intestine, Small/drug effects , Intestine, Small/immunology
4.
Annu Rev Virol ; 4(1): 369-391, 2017 09 29.
Article in English | MEDLINE | ID: mdl-28715972

ABSTRACT

α, ß, and θ defensins are effectors of the innate immune system with potent antibacterial, antiviral, and antifungal activity. Defensins have direct antiviral activity in cell culture, with varied mechanisms for individual viruses, although some common themes have emerged. In addition, defensins have potent immunomodulatory activity that can alter innate and adaptive immune responses to viral infection. In some cases, there is evidence for paradoxical escape from defensin neutralization or enhancement of viral infection. The direct and indirect activities of defensins have led to their development as therapeutics and vaccine components. The major area of investigation that continues to lag is the connection between the effects of defensins in cell culture models and viral pathogenesis in vivo. Model systems to study defensin biology, including more physiologic models designed to bridge this gap, are also discussed.


Subject(s)
Defensins/metabolism , Virus Diseases/immunology , alpha-Defensins/metabolism , beta-Defensins/metabolism , Adenoviridae/drug effects , Adenoviridae/pathogenicity , Animals , Antiviral Agents/pharmacology , Defensins/genetics , Defensins/pharmacology , Defensins/therapeutic use , HIV/drug effects , HIV/pathogenicity , Herpesviridae/drug effects , Herpesviridae/pathogenicity , Humans , Immunity, Innate , Immunomodulation , Mice , Papillomaviridae/drug effects , Papillomaviridae/pathogenicity , Virus Diseases/drug therapy , alpha-Defensins/genetics , alpha-Defensins/pharmacology , alpha-Defensins/therapeutic use , beta-Defensins/genetics , beta-Defensins/pharmacology , beta-Defensins/therapeutic use
5.
PLoS Pathog ; 13(6): e1006446, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28622386

ABSTRACT

The small intestinal epithelium produces numerous antimicrobial peptides and proteins, including abundant enteric α-defensins. Although they most commonly function as potent antivirals in cell culture, enteric α-defensins have also been shown to enhance some viral infections in vitro. Efforts to determine the physiologic relevance of enhanced infection have been limited by the absence of a suitable cell culture system. To address this issue, here we use primary stem cell-derived small intestinal enteroids to examine the impact of naturally secreted α-defensins on infection by the enteric mouse pathogen, mouse adenovirus 2 (MAdV-2). MAdV-2 infection was increased when enteroids were inoculated across an α-defensin gradient in a manner that mimics oral infection but not when α-defensin levels were absent or bypassed through other routes of inoculation. This increased infection was a result of receptor-independent binding of virus to the cell surface. The enteroid experiments accurately predicted increased MAdV-2 shedding in the feces of wild type mice compared to mice lacking functional α-defensins. Thus, our studies have shown that viral infection enhanced by enteric α-defensins may reflect the evolution of some viruses to utilize these host proteins to promote their own infection.


Subject(s)
Adenoviridae Infections/virology , Adenoviridae/physiology , Intestine, Small/metabolism , alpha-Defensins/metabolism , Adenoviridae/genetics , Animals , Female , Host-Pathogen Interactions , Humans , Intestine, Small/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Virus Shedding , alpha-Defensins/genetics
6.
J Virol ; 90(11): 5216-5218, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27009960

ABSTRACT

Defensins are innate immune effector peptides expressed at mucosal surfaces throughout the human body and are potently antiviral in vitro The role of defensins in viral pathogenesis in vivo is poorly understood; however, recent studies have revealed that defensin-virus interactions in vivo are complicated and distinct from their proposed antiviral mechanisms in vitro These findings highlight the need for additional research that connects defensin neutralization of viruses in cell culture to in vivo antiviral mechanisms.


Subject(s)
Defensins/metabolism , Immunomodulation , Mucous Membrane/immunology , Mucous Membrane/virology , Virus Diseases/immunology , Viruses/immunology , Animals , Antiviral Agents/immunology , Antiviral Agents/metabolism , Defensins/immunology , Humans , Mucous Membrane/chemistry , Mucous Membrane/physiology , Virus Diseases/virology , Viruses/metabolism , alpha-Defensins/immunology , alpha-Defensins/metabolism
7.
Appl Environ Microbiol ; 80(3): 959-71, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24271174

ABSTRACT

We present the results of a study using high-throughput whole-transcriptome sequencing (RNA-seq) and vibrational spectroscopy to characterize and fingerprint pathogenic-bacterium injury under conditions of unfavorable stress. Two garlic-derived organosulfur compounds were found to be highly effective antimicrobial compounds against Cronobacter sakazakii, a leading pathogen associated with invasive infection of infants and causing meningitis, necrotizing entercolitis, and bacteremia. RNA-seq shows changes in gene expression patterns and transcriptomic response, while confocal micro-Raman spectroscopy characterizes macromolecular changes in the bacterial cell resulting from this chemical stress. RNA-seq analyses showed that the bacterial response to ajoene differed from the response to diallyl sulfide. Specifically, ajoene caused downregulation of motility-related genes, while diallyl sulfide treatment caused an increased expression of cell wall synthesis genes. Confocal micro-Raman spectroscopy revealed that the two compounds appear to have the same phase I antimicrobial mechanism of binding to thiol-containing proteins/enzymes in bacterial cells generating a disulfide stretching band but different phase II antimicrobial mechanisms, showing alterations in the secondary structures of proteins in two different ways. Diallyl sulfide primarily altered the α-helix and ß-sheet, as reflected in changes in amide I, while ajoene altered the structures containing phenylalanine and tyrosine. Bayesian probability analysis validated the ability of principal component analysis to differentiate treated and control C. sakazakii cells. Scanning electron microscopy confirmed cell injury, showing significant morphological variations in cells following treatments by these two compounds. Findings from this study aid in the development of effective intervention strategies to reduce the risk of C. sakazakii contamination in the food production environment and on food contact surfaces, reducing the risks to susceptible consumers.


Subject(s)
Allyl Compounds/pharmacology , Anti-Bacterial Agents/pharmacology , Cronobacter sakazakii/drug effects , Disulfides/pharmacology , Garlic/chemistry , Spectrum Analysis, Raman , Sulfides/pharmacology , Transcriptome , Allyl Compounds/isolation & purification , Anti-Bacterial Agents/isolation & purification , Cronobacter sakazakii/ultrastructure , Disulfides/isolation & purification , Microscopy, Electron, Scanning , Protein Conformation/drug effects , Sulfides/isolation & purification , Sulfoxides
SELECTION OF CITATIONS
SEARCH DETAIL
...